• Aucun résultat trouvé

Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up

N/A
N/A
Protected

Academic year: 2021

Partager "Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up"

Copied!
5
0
0

Texte intégral

(1)

Locally recurrent or metastatic breast cancer: ESMO

Clinical Practice Guidelines for diagnosis, treatment and

follow-up

F. Cardoso

1

, E. Senkus-Konefka

2

, L. Fallowfield

3

, A. Costa

4

& M. Castiglione

5

On behalf of the ESMO Guidelines Working Group*

1

Department of Medical Oncology, Jules Bordet Institute, Brussels, Belgium;2

Department of Oncology and Radiotherapy, Medical University of Gdan´sk, Gdan´sk, Poland; 3

Brighton and Sussex Medical School, University of Sussex, UK;4

European School of Oncology, Milan and Maugeri Foundation Breast Unit, Pavia, Italy;5 RGT, University of Geneva, Geneva, Switzerland

incidence

Breast cancer is the most common cancer in women in many countries, including developing countries. In 2006, the crude incidence in the European Union was 109.8/100 000, the mortality was 38.4/100 000 women/year. Since 1990 the incidence rate has increased 1.5% annually. Due to advances both in early detection and in adjuvant treatment, mortality rates from breast cancer have been decreasing steadily in most western countries since the early 1990s. However, it is still the leading cause of cancer mortality in women. Approximately 6% of breast cancers are metastatic at diagnosis with a 5-year survival rate of 21%. Depending on prognostic factors, in the worst scenario, up to 30% of node-negative and up to 70% of node-positive breast cancers will relapse. The prevalence of metastatic disease is high because many women live with the disease for several years.

diagnosis

Clinical suspicion must be confirmed by imaging (including functional imaging); additional information may be provided by laboratory tests.

Efforts should be made to obtain histopathological confirmation particularly in the situation of an isolated metastatic lesion. Biological markers important for treatment decisions, such as hormonal receptors and HER2 status, should be evaluated in the metastatic lesion whenever possible. Potential exceptions are (a) situations where the biopsy procedure is too risky, (b) the time elapsed between the primary tumour and the metastatic disease diagnosis is relatively short or (c) when the results of the biopsy are unlikely

to change the therapeutic attitude (e.g. pre-existent

contraindications for the use of chemotherapy or anti-HER2 therapies).

There is no proven value of routine diagnostic tests ‘screening’ for metastatic disease in asymptomatic patients. However, the available data are from a time when neither biological therapy nor efficacious (in terms of local control) and less invasive loco-regional techniques, such as radiosurgery for central nervous system (CNS) metastases or radiofrequency for liver metastases, were available. Additionally, new techniques are now available, such as magnetic resonance imaging (MRI), PET scan, PET–CT, circulating tumour cells and others, that may allow for the detection of very early metastatic disease. Therefore, new studies are needed to evaluate the role of early diagnosis of metastatic disease in this new context.

The occurrence of loco-regional recurrence is often associated with distant spread and such patients should undergo full staging procedures before undergoing local treatments.

staging and risk assessment

 Complete history, including:

(i) menopausal status and co-morbidities;

(ii) detailed history of the primary tumour, its biology, management and status at last follow-up;

(iii) history of recurrent/metastatic disease including duration, previous sites of involvement, previous treatments and their effect;

(iv) current symptoms, performance status, socio-economic background and preferences (Table 1).

 Detailed physical examination.

 Blood tests: complete blood count, liver and renal function

tests, alkaline phosphatase, calcium and, if applicable, specific tests required for particular treatments such as urinary protein. The clinical value of tumour markers has not been well proven; however, their use may be of value to assess the

*Correspondence to: ESMO Guidelines Working Group, ESMO Head Office, Via L. Taddei 4, CH-6962 Viganello-Lugano, Switzerland;

E-mail: clinicalrecommendations@esmo.org

Approved by the ESMO Guidelines Working Group: August 2003, last update February 2010. This publication supercedes the previously published version—Ann Oncol 2009; 20 (Suppl 4): iv15–iv18.

Conflict of interest: The authors have reported no conflicts of interest.

(2)

efficacy of treatment particularly in patients with non-measurable disease.

 Chest X-ray or computed tomography (CT), abdominal

ultrasound, CT or MRI should be used to identify visceral disease.

 Bone scintigraphy, with confirmation of lesions by X-ray/CT/

MRI.

 CT and/or MRI of the CNS should be symptom driven.  PET/PET–CT may be useful for identifying the site of relapse,

particularly when traditional imaging methods are equivocal or conflicting. It may also be helpful to identify an isolated metastatic lesion, since this subset of patients may benefit from a more aggressive multidisciplinary approach.

 Estrogen and progesterone receptors, HER2 receptors and

proliferation markers of the metastatic lesion should be obtained, if possible, and particularly if not available on the primary tumour.

 Cardiac assessments, in particular in HER2(+) patients.  Circulating tumour cells is still an experimental technique

and should not be used outside a clinical trial.

treatment

local–regional recurrence

Isolated local–regional recurrence should be treated like a new primary with a curative intent. If feasible, complete excision of recurrent tumour is recommended. In patients not exposed to postoperative irradiation, radical radiotherapy to chest wall and (in most cases) regional lymph node areas should be given. In those previously irradiated, the value of re-irradiation is not proven; however, re-re-irradiation to limited areas in the chest wall may be applied, after a careful benefit– risk balance, taking into consideration the duration of radiation-free period, intensity of post-radiotherapy changes and the risk of local–regional relapse. Inoperable patients can, if feasible, undergo radical radiotherapy to chest wall and regional lymph node areas with boost to macroscopic disease sites. However, in these patients, primary systemic therapy to

decrease the size of the tumour and render it operable should be the first choice.

The value of ‘secondary or pseudo-adjuvant’ systemic treatment is not well proven. The role of ‘secondary or pseudo-adjuvant’ chemotherapy is a subject of ongoing randomized studies [II, B].

metastatic disease

 The management of metastatic breast cancer (MBC) should

involve all appropriate specialties in a multi/interdisciplinary team (medical, radiation, surgical and imaging oncologists, palliative care specialist, psychosocial support), and patients should be offered personalized appropriate psychosocial, supportive and symptom-related interventions as a routine part of their care.

 There are few proven standards of care in MBC management,

therefore well-designed, independent, prospective randomized trials are a priority.

 The vast majority of MBC is incurable and hence the main

treatment goal is palliation, with the aim of maintaining/ improving quality of life, and possibly improving survival.

 The realistic treatment goals should be discussed with the

patient and her family from the beginning and the patient should be encouraged to actively participate in all decisions. Patients’ preferences should always be taken into account.

 Co-ordination and continuity of care may need to be

facilitated by a specialist breast care nurse or key worker.

 Systemic treatment options for MBC are endocrine therapy,

chemotherapy and biological agents such as trastuzumab, bevacizumab and lapatinib [I, A] (see Table 1).

 The choice of therapy should be made after consideration of

factors such as: previous therapies and response to them, disease-free interval, endocrine responsiveness, HER2 status, tumour burden (defined as number and site of metastases), menopausal status, biological age and co-morbidities (including organ dysfunction), performance status, need for rapid disease/symptom control, socio-economic and psychological factors, patient’s preference and available therapies in the patient’s country.

 Patients’ preferences should always be taken into account not

only about treatment options but also methods of treatment administration (i.v. or oral).

 For the majority of patients, overall survival outcomes from

sequential use of single cytotoxic drugs are equivalent to combination chemotherapy. The choice between both these options should primarily take into account the need for a rapid and significant response and quality of life.

 Duration of each regimen and number of regimens should be

tailored to each individual patient.

 Radiation therapy is an integral part of palliative treatment.

The most common indications for palliative radiotherapy include:

 Bone metastases that are painful or carry a risk of fractures

and/or neurological complications (radiotherapy options include ‘limited field’ external beam irradiation, hemi-body irradiation and application of radioactive ‘bone-seeking’ isotopes);

Table 1. Factors to consider in risk assessment and treatment decision making for MBC

Disease-related factors Patient-related factors

Disease-free interval Patient’s preferences

Previous therapies and response

Biological age Biological factors (hormonal

receptors, HER2)

Menopausal status Tumour burden (number

and site of metastases)

Co-morbidities and performance status Need for rapid disease/

symptom control

Socio-economic and psychological factors Available therapies in the

(3)

 Brain metastases (several series have shown that, in

patients with single or a few metastatic foci, stereotactic radiosurgery can be used with equally good local control and less side-effects than whole brain radiotherapy);

 Painful or fungating soft tissue masses.

 For limited metastatic presentations surgery or radical

radiotherapy may be considered.

 Bisphosphonates should be used for the treatment of

hypercalcaemia and clinically evident bone metastases (to palliate symptoms and decrease risk of bone events) [I, A]. The timing and optimal duration of bisphosphonates are unknown. The choice of drugs, their timing, optimal duration, methods of administration and side-effects have significant consequences for a patient’s life-style especially in terms of their ability to adhere to treatment. Monitoring of acceptability and adherence is crucial, and choice where possible should be offered.

patients with luminal-type breast

cancer (hormone receptor-positive

breast cancer, irrespective of HER2

status)

 Endocrine therapy is the preferred option except when

clinically aggressive disease mandates a quicker response or if there are doubts regarding the endocrine responsiveness of the tumour. Available endocrine agents are listed in Table 2.

 The choice of endocrine agent should be individualized

according to the safety profile, patient’s co-morbidities and tumour biology.

 Apart from combination of tamoxifen with ovarian

suppression in premenopausal patients there is no rationale for the use of combination hormonal therapies.

 The value of maintenance with hormonal treatment after

chemotherapy has not been confirmed by controlled clinical studies, but is a reasonable approach.

 Concomitant chemo-hormonal therapy is discouraged.

 In the case of HER2 overexpression/amplification, addition of

anti-HER2 therapies to hormonal treatment is beneficial.

premenopausal patients

If no prior adjuvant tamoxifen or if discontinued for >12 months: tamoxifen with ovarian ablation (luteinizing hormone releasing hormone analogue or surgery) is the preferred option [I, B]. Otherwise, third-generation aromatase inhibitors may be considered after or concomitantly with ovarian ablation. Further treatment lines (in patients who had undergone ovarian ablation/suppression) do not differ from those used in the postmenopausal population (as described below).

postmenopausal patients

If no prior adjuvant third-generation aromatase inhibitors (anastrozole, letrozole, exemestane) or if discontinued for >12 months these are the preferred options since they have consistently shown superior results to tamoxifen as first-line therapy in terms of response rate, time to progression and, for letrozole, in 2-year overall survival [II, A]. Caution should be given to the risk of accelerated bone loss in these patients.

Tamoxifen remains an acceptable first-line therapy. Patients given tamoxifen should be instructed to avoid use of drugs modulating the activity of CYP2D6, such as some selective serotonin reuptake inhibitor antidepressants (paroxetine, fluoxetine).

Second-line hormone therapy may include tamoxifen, third-generation aromatase inhibitors (if not previously used), fulvestrant, megestrol acetate and androgens. No definitive recommendation can be given for endocrine treatment cascade, and particularly, the best option after progression on a third-generation aromatase inhibitor is currently unknown.

Patients with clear evidence of endocrine resistance should be offered chemotherapy or participation in clinical trials.

patients with ‘triple negative’ breast

cancer (hormone receptor-negative and

HER2-non-overexpressed/

non-amplified breast cancer)

 Patients having hormone receptor-negative tumours are

candidates for cytotoxic chemotherapy. Available agents/ regimens are listed in Table 3.

 The only standard of care with level 1 evidence is the use of

a taxane-based regimen as first-line therapy in patients progressing after adjuvant anthracycline-based chemotherapy [I A]. The selection of the best agent/regimen should be individualized and should take into account the factors listed in Table 1.

 For the majority of patients, overall survival outcomes from

the sequential use of single cytotoxic drugs are equivalent to that of combination chemotherapy, with less associated toxicity and better quality of life. Therefore, in the absence of the need for a rapid and significant response for symptom control or life-threatening disease, preference should be given to the sequential use of single cytotoxic agents. However, very few randomized clinical trials have correctly addressed this question

Table 2. Available endocrine therapies for MBC

Class of agent

Selective estrogen receptor modulators

Tamoxifen; toremifene Estrogen receptor down-regulator Fulvestrant

Luteinizing hormone-releasing hormone analogues

Goserelin; leuprorelin Third-generation aromatase

inhibitors

Non-steroidal Anastrozole, letrozole

Steroidal Exemestane

Progestins Medroxyprogesterone acetate;

megestrol acetate Androgens

(4)

and there is an urgent need for a well-designed, prospective randomized trial to compare sequential single-agent with combination chemotherapy as first-line therapy of MBC.

 There is no standard approach for patients requiring

second-or further-line treatment since there are no data suppsecond-orting the superiority of any particular regimen.

 Duration of each regimen and number of regimens should be

tailored to each individual patient. Continuing beyond third-line may be justified in patients with good performance status and response to previous chemotherapy.

 High-dose chemotherapy should not be proposed.

 The role of bevacizumab will be discussed in the section on

other biological agents.

patients with HER2-positive

(overexpressed/amplified) breast

cancer

 Patients should be treated with trastuzumab with or without

chemotherapy [II, B].

 Trastuzumab should be offered early to all HER2-positive

MBC patients.

 Cardiac monitoring should be performed before and while on

trastuzumab therapy.

 The bulk of retrospective data and results of the phase III

randomized Trial Beyond Progression show that continuing trastuzumab after the first disease progression, associated

with a different chemotherapy regimen is superior to the discontinuation of this agent. With the approval of lapatinib for the treatment of MBC, the question of continuing trastuzumab or changing to lapatinib at the time of first progression remains open.

 Lapatinib has shown a significant increase in time to

progression in combination with capecitabine in patients progressing after trastuzumab.

 Addition of anti-HER2 agents (trastuzumab and lapatinib) to

endocrine therapy allows for prolongation of progression-free survival (PFS) and may be a viable option in patients with ER/PgR-positive and HER2-positive tumours.

 Other anti-HER2 or pan-anti-HER agents, such as

pertuzumab and HKI-272, are currently under investigation as are combinations of trastuzumab with other biological agents with or without chemotherapy to tackle the problem of resistance to trastuzumab.

other biological agents

 Bevacizumab, an anti-angiogenic agent, has been approved by

the FDA and the EMEA for use in combination with paclitaxel as first-line treatment of MBC after showing a benefit of 6 months in PFS in the ECOG 2100 study. However, in two other randomized phase III trials, the AVADO and RIBBON studies, the benefit of bevacizumab in an unselected breast cancer population was of 1 month in PFS with no significant benefit in overall survival. Efforts must continue to be made to clearly identify which patients may benefit from this expensive therapy.

 Several biological or targeted agents are currently under

active investigation as single agents or in combination.

response evaluation

 Response evaluation is routinely recommended after 2–3

months of endocrine therapy and after two or three cycles of chemotherapy by clinical evaluation, subjective symptom evaluation, blood tests and repeating the initially

abnormal radiological examinations with comparative measurements. However, the interval between assessments should be tailored to the clinical needs of the patient and to the aggressiveness of the disease. In the case of clinical suspicion of progressive disease appropriate tests (imaging and laboratory) should be performed irrespective of scheduled examinations, if necessary including areas not imaged in previous tests.

 Serum tumour markers (CA 15-3) may be helpful in

monitoring response, particularly in the case of not easily measurable disease, but should not be used as the only determinant for treatment decision.

 The role of PET/PET–CT in response assessment is still under

investigation.

 Maintenance of a good quality of life is paramount and can

best be achieved with prompt amelioration of symptoms and side-effects of treatment. Psychometrically sound, well-validated questionnaires are available to measure

patient-Table 3. Selection of available chemotherapy agents/regimens for MBC

Non-anthracycline-containing

Cyclophosphamide/methotrexate/5-fluorouracil (CMF) Platinum-based combinations (e.g. cisplatin + 5-fluorouracil) Capecitabine

Vinorelbine Gemcitabine

Capecitabine + vinorelbine Vinorelbine 6 gemcitabine

Oral cyclophosphamide with or without methotrexate (metronomic chemotherapy)

Anthracycline-containing

Doxorubicin or epirubicin monotherapy (weekly or 3-weekly) Doxorubicin or epirubicin/cyclophosphamide

Liposomal doxorubicin with or without cyclophosphamide 5-fluorouracil/doxorubicin/cyclophosphamide

5-fluorouracil/epirubicin/cyclophosphamide Taxane-containing

Paclitaxel monotherapy weekly

Docetaxel monotherapy 3-weekly or weekly Doxorubicin/taxane (paclitaxel or docetaxel) Epirubicin/taxane (paclitaxel or docetaxel) Docetaxel/capecitabine

Paclitaxel/gemcitabine Paclitaxel/vinorelbine Paclitaxel/carboplatin New cytotoxic agents

Ixabepilone

(5)

reported outcomes. These should be employed regularly to help assess the impact of treatment and to monitor symptoms that demand supportive intervention promptly.

follow-up

 Follow-up after the treatment of local–regional recurrence

may be carried out as for primary breast cancer.

 Patients with MBC must be seen frequently enough to

provide best possible palliation of symptoms and quality of life, which means on average every 2–3 months if on endocrine therapy and every one or two cycles of chemotherapy.

 Finally patients need good quality information and a care plan

outlining all aspects of treatment and care, clarification of the purpose of different treatments, their side-effects and potential impact on functional, emotional and social well-being.

note

Levels of evidence [I–V] and grades of recommendation [A–D] as used by the American Society of Clinical Oncology are given in square brackets. Statements without grading were considered justified standard clinical practice by the experts and the ESMO faculty.

literature

1. ESO-MBC Task Force. Metastatic breast cancer. Recommendations proposal from the European School of Oncology (ESO)-MBC Task Force. Breast 2007; 16: 9–10.

2. Kataja V, Castiglione M. Locally recurrent or metastatic breast cancer: ESMO Clinical Recommendations for diagnosis, treatment and follow-up. Ann Oncol 2008; 19 (Suppl 2): ii11–ii13.

3. Network NCC. 2008. NCCN Clinical Practice Guidelines in Oncology: Breast Cancer, Version 2. In Edition 2008.

4. Colozza M, de Azambuja E, Personeni N et al. Achievements in systemic therapies in the pre-genomic era in metastatic breast cancer. Oncologist 2007; 12: 253–270.

5. Cardoso F, Bedard PL et al. on behalf of the ESO-MBC Task Force. International guidelines for management of metastatic breast cancer: combination vs sequential single-agent chemotherapy. J Natl Cancer Inst 2009; 101: 1174–1181.

6. Chan S, Friedrichs K, Noel D et al. Prospective randomized trial of docetaxel versus doxorubicin in patients with metastatic breast cancer. J Clin Oncol 1999; 17: 2341–2354.

7. O’Shaughnessy J, Miles D, Vukelja S et al. Superior survival with capecitabine plus docetaxel combination therapy in anthracycline-pretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 2002; 20: 2812–2823.

8. Albain KS, Nag SM, Calderillo-Ruiz G et al. Gemcitabine plus paclitaxel versus paclitaxel monotherapy in patients with metastatic breast cancer and prior anthracycline treatment: final results of a global phase III study. J Clin Oncol 2008; 26: 3950–3957.

9. Burstein HJ, Kuter I, Campos SM et al. Clinical activity of trastuzumab and vinorelbine in women with HER2-overexpressing metastatic breast cancer. J Clin Oncol 2001; 19: 2722–2730.

10. Miller K, Wang M, Gralow J et al. Paclitaxel pus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 2007; 357: 2666–2676. 11. Gennari A, Amadori D, De Lena M et al. Lack of benefit of maintenance paclitaxel

in first-line chemotherapy in metastatic breast cancer. J Clin Oncol 2006; 24: 3912–3918.

12. Ghersi D, Wilcken N, Simes J et al. Taxane containing regimens for metastatic breast cancer. Cochrane Database Syst Rev 2003; issue 3: CD003366. 13. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in

solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45: 228–247.

14. Slamon DJ, Leyland-Jones B et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344: 783–792.

15. Marty M, Cognetti F, Maraninchi D et al. Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J Clin Oncol 2005; 23: 4265–4274.

16. Geyer CE, Forster J, Lindquist D et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006; 355: 2733–2743. 17. Mackey JR, Kaufman B, Clemens M et al. Trastuzumab prolongs

progression-free survival in hormone-dependent and HER2- positive metastatic breast cancer. Breast Cancer Res Treat 2006; 100 (Suppl 1): S5.

18. Johnston S, Pippen J Jr, Pivot X et al. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor–positive metastatic breast cancer. J Clin Oncol 2009; 27: 5538–5546. 19. Gibson L, Dawson C, Lawrence D, Bliss J. Aromatase inhibitors for treatment of advanced breast cancer in postmenopausal women. Cochrane Database Syst Rev 2007; issue 1: CD003370.

20. Costelloe CM, Rohren EM, Madewell JE et al. Imaging bone metastases in breast cancer: techniques and recommendations for diagnosis. Lancet Oncol 2009; 10: 606–614.

Figure

Table 1. Factors to consider in risk assessment and treatment decision making for MBC
Table 2. Available endocrine therapies for MBC
Table 3. Selection of available chemotherapy agents/regimens for MBC

Références

Documents relatifs

More generally, on the basis of this and other experiments, we must conclude that Maxwell’s equations are correct and that the speed of electromagnetic radiation is the same in

Par suite le point I est un point d’inflexion de C.. 3) L’allure du nuage est proche de l’allure de (C) et n’a pas l’allure d’une droite alors le deuxième ajustement est

Mass spectrometry data of Arabidopsis thaliana proteins with the strongest association to the RxLR24 effector after co-immunoprecipitation..

Analysis of protein-protein interactions of human ARMC6 protein by co-immunoprecipitation The bait construct TRF2/pGBKT7-DEST was expressed in 25 µl of a RRL, the prey constructs

Dans une analyse des besoins sur les formations en lien avec l’islam menée par le Centre Suisse Islam et Société (CSIS) de l’Université de Fribourg (2016), des experts de

Tous les deux, vous n'avez peur de rien, vous sautez de branche en branche et vous ne voyez pas le danger.. Parfois, vous faites de longues promenades puis vous revenez auprès de

 Dictée préparée n°3 - L’âne portant du sel (étape : correction) Vendre... Quand il est au plus haut dans le ciel, il est midi,

Il s'agit d'une sorte de calendrier rustique vantant le travail agricole, les rites religieux et les saisons.. Cette scène représenté la cueillette des pommes: un personnage est